Weekly Digests
‹ Back to September

Rebooting the cancer immunity cycle by inducing necroptosis

September 19, 2018

The cancer immunity cycle is the body’s natural way of staving off cancer, but in instances when cancer has already escaped this cycle, inducing immunogenic tumor cell death may be an effective way to reboot the system. In a recent study published in Nature Communications, Van Hoecke et al. explored the use of intratumorally delivered mRNA encoding the necroptotic cell death executioner protein MLKL (MLKL-mRNA) to induce necroptosis and a subsequent antitumor immune response.

To begin, Van Hoecke et al. generated and delivered mRNA encoding MLKL, tBid (an inducer of apoptosis), or Fluc (an irrelevant reporter) to B16 melanoma cells. The MLKL-mRNA induced necroptosis (marked by a lack of caspase activity or caspase-3 processing, followed by cell rounding, swelling, and membrane permeabilization), while tBid induced apoptosis (marked by blebbing, caspase activity, cleavage fragments of caspase-3, and loss of membrane integrity). MLKL-mRNA and tBid-mRNA both induced similar levels of tumor cell death, which was a three-fold increase over the delivery of control mRNA or saline.

In mice bearing B16 melanoma, intratumorally delivered mRNA was taken up by tumor cells, with peak protein expression occurring 12 hours after electroporation. In both B16-OVA melanoma and CT26-OVA colon carcinoma models, tBid-mRNA delayed tumor growth and increased median survival. MLKL-mRNA delayed tumor growth more significantly and improved survival further. MLKL-mRNA also outperformed repeated treatments of doxorubicin, a known apoptosis inducer, with fewer negative side effects.

Since in vitro results had shown that tBid-mRNA and MLKL-mRNA induced similar levels of tumor cell death, the researchers suspected that the difference in antitumor efficacy might be due to participation by the immune system following the release of damage-associated molecular patterns (DAMPs) and tumor antigens. To test whether there was a systemic immune effect occurring, the researchers treated tumor-bearing mice, then removed the primary tumor and rechallenged mice in the same location. They found that treatment with MLKL-mRNA protected 40% of B16 melanoma-bearing mice and 100% of CT26 colon carcinoma-bearing mice. Similarly, the researchers tested for an abscopal effect by implanting a primary tumor on one side, and later, a secondary tumor on the opposite side. Treatment of the primary tumor with MLKL-mRNA significantly delayed the growth of both primary and secondary tumors, indicating a systemic immune effect; this effect was weak in tBid-mRNA-treated mice and not observed in controls.

To further investigate the immune response involved in the antitumor efficacy of MLKL-mRNA, the researchers moved to determine whether the MLKL-induced necroptosis primes T cell responses. To this end, the researchers adoptively transferred reporter-labeled, OVA-specific CD4+ and CD8+ T cells into mice bearing B16-OVA melanoma. When they removed the lymph nodes of MLKL-mRNA-treated animals, they found evidence of T cell proliferation and increased numbers of OVA-specific CD4+ and CD8+ T cells. An in vivo killing assay showed that MLKL-mRNA increased specific killing to 75% of target cells, while tBid-mRNA induced only 40% target cell killing, and controls induced no significant target cell lysis. Further, the researchers were able to identify T cells from MLKL-mRNA-treated mice that were specific to known neoepitopes in both B16 melanoma and CT26 colon carcinoma, but not the wild-type proteins. These neoepitope-specific responses were not identified in mice treated with tBid-mRNA or controls.

With evidence for the participation of T cells in the antitumor efficacy of MLKL-mRNA, the researchers next explored the mechanism by which the T cells become activated. Following MLKL-mRNA treatment, the researchers observed an influx of cDC1 (Batf3-dependent) and cDC2 (IRF4-dependent) dendritic cells into the tumor bed and tumor draining lymph nodes. Various knockout models revealed that the presence of cDC1 dendritic cells, type I interferon signaling, and migration of dendritic cells to the draining lymph node were were all required in order to mount T cell priming and a subsequent OVA-specific cytotoxic immune response.

Because intratumoral treatment with MLKL-mRNA increases T cell priming and infiltration into the tumor, Van Hoeke et al. hypothesized that their treatment would synergize with anti-PD-1 blockade, which would overcome T cell suppression in the tumor. They tested their hypothesis in mice with primary and abscopal tumors. Treatment of the primary tumor with MLKL-mRNA combined with anti-PD-1 treatment led to significantly improved tumor suppression in both the primary tumor and the distant untreated tumor, and improved survival compared to either monotherapy.

To get a better idea of whether this therapy could translate to the clinic, the researchers assessed mRNA encoding for human MLKL and found that it could effectively kill human cancer cells in vitro. In a humanized mouse model bearing human follicular lymphoma tumors, hMLKL-mRNA induced a strong antitumor effect that significantly delayed tumor growth and increased median survival, outperforming tBid-mRNA treatment and controls. While the preclinical data is promising, further preclinical research is necessary to fully understand the mechanism of MLKL-induced necroptosis. Clinical translation would also come with a host of additional hurdles, including the accessibility of tumors and their larger size. Still, evidence that MLKL-encoding mRNA can act as an in situ vaccine presents an exciting new avenue for immunotherapy research.

by Lauren Hitchings

References:

Van Hoecke L., Van Lint S., Roose K., Van Parys A., Vandenabeele P., Grooten J.,Tavernier J., De Koker S., Saelens X. Treatment with mRNA coding for the necroptosis mediator MLKL induces antitumor immunity directed against neo-epitopes. Nat Commun. 2018 Aug 24.

In the Spotlight...

An RNA toolbox for cancer immunotherapy

RNA is a multifunctional molecule with diverse capabilities including protein coding, intracellular regulation, structural scaffolding, and induction of innate immune reactions. Pastor et al. review how these diverse functional properties can be utilized for immunotherapeutic applications. Although only one RNA-based therapeutic is an FDA approved drug (an anti-VEGF antagonist), many are in clinical development. Key opportunities are low cost due to simple, rapid chemical synthesis, readily modifiable nucleotides, and short half-life (in case of unanticipated immune toxicities); key challenges are delivery and stability.

A homing system targets therapeutic T cells to brain cancer

Samaha et al. designed a brain tumor homing system (HS) by engineering T cells to express a multimerized version of the D3 domain of the ALCAM-binding CD6 exodomain together with CD6 transmembrane and signaling endodomains. In a two-step process, HS T cells bind to ALCAM, which is overexpressed on brain tumor endothelial cells, sensitizing the T cells to the downregulated levels of ICAM1 in tumor endothelium and allowing for infiltration of T cells into the tumor. In mice, HER2-targeting CAR 5HS T cells selectively infiltrated the GBM tumor, induced tumor regression, and increased survival compared with HER2 CAR T cells and normal T cells.

SD-101 in Combination with Pembrolizumab in Advanced Melanoma: Results of a Phase 1b, Multicenter Study

In an ongoing phase 1b dose escalation study, 22 patients with stage IIIC/IV metastatic melanoma were treated with intratumoral SD-101 (a CpG-oligonucleotide that stimulates TLR9) in combination with systemic pembrolizumab. Most adverse events were grade 1 or 2 and transient. In the 9 anti-PD-1/PD-L1 naive patients, ORR was 78% (2 patients with CR, 5 with PR), with both injected and non-injected lesions responding. Out of the 13 patients previously treated with anti-PD-1/PD-L1, 2 patients had a PR, and the rest went on to develop progressive disease. The treatment increased IFNα responses and infiltration of CD8+ T cells into the tumor.

In Situ Vaccination with a TLR 9 Agonist and Local Low Dose Radiation Induces Systemic Responses in Untreated Indolent Lymphoma

In a phase 1/2 dose escalation study, 29 previously untreated patients with low-grade B-cell lymphoma were treated with SD-101 (a CpG-oligonucleotide) and low-dose radiation at the same, single tumor site. 8 patients had grade 3 adverse events, primarily flu-like symptoms. Tumor burden was reduced in 26 patients (with 1 CR and 7 PR) and 24 patients had response in non-treated lesions. Responses were durable. The treatment increased IFNα response, CD8+, and CD4+ effector T cells, and decreased TFH and Tregs in the tumor. Low pretreatment levels of Tregs, Ki67+ CD8+ T cells, and granzyme B+ CD8+ T cells were associated with better clinical outcomes.

First-In-Human Phase I Trial of a Tumor-Targeted Cytokine (NHS-IL12) in Subjects with Metastatic Solid Tumors

In a phase 1 trial, 59 patients with metastatic or locally advanced solid tumors were treated subcutaneously with NHS-IL12, an immunocytokine composed of two IL-12 heterodimers fused to an antibody that targets histones in areas of tumor necrosis. Treatment was generally well tolerated up to the dose of 16.8 μg/kg. The best response was stable disease in 15 patients. In peripheral blood, IFNγ, IL-10, and mature and activated NK and NKT cells increased, and CD4+ EMRA T cells and plasmacytoid DCs decreased. In the tumor, there was an increase in TCR diversity and TIL density in patients with high IFNγ response.

Decreased Suppression and Increased Phosphorylated STAT3 in Regulatory T-cells are Associated with Benefit from Adjuvant PD-1 Blockade in Resected Metastatic Melanoma

Woods et al. demonstrated that treatment of resected high-risk melanoma with adjuvant anti-PD-1 for 13 weeks increased the number of circulating Tregs with reduced suppressive function and increased pSTAT3 expression in non-relapsing compared to relapsing patients. Ex vivo studies further showed that increased proliferation of Tregs was due to upregulated production of IL-10 by CD8+ T cells via the STAT3 pathway and conversion of conventional CD4+ T cells to inducible Tregs. In metastatic patients treated with anti-PD-1, increased survival positively correlated with increased pSTAT3 expression in Tregs and CD8+ T cells.

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.